EP4181872A1 - Composition pharmaceutique aqueuse comprenant un antagoniste du récepteur p2y12 - Google Patents

Composition pharmaceutique aqueuse comprenant un antagoniste du récepteur p2y12

Info

Publication number
EP4181872A1
EP4181872A1 EP21739730.6A EP21739730A EP4181872A1 EP 4181872 A1 EP4181872 A1 EP 4181872A1 EP 21739730 A EP21739730 A EP 21739730A EP 4181872 A1 EP4181872 A1 EP 4181872A1
Authority
EP
European Patent Office
Prior art keywords
pharmaceutical composition
aqueous pharmaceutical
buffer
composition according
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21739730.6A
Other languages
German (de)
English (en)
Inventor
Stephan Buchmann
Amandine FRAICHARD
Charlyse HERRMANN
Céline LIENHART
Markus Von Raumer
Tobias WERK
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Idorsia Pharmaceuticals Ltd
Original Assignee
Idorsia Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Idorsia Pharmaceuticals Ltd filed Critical Idorsia Pharmaceuticals Ltd
Publication of EP4181872A1 publication Critical patent/EP4181872A1/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/186Quaternary ammonium compounds, e.g. benzalkonium chloride or cetrimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors

Definitions

  • Aqueous Pharmaceutical Composition comprising a P2Y12 Receptor Antagonist
  • the present invention relates to an aqueous pharmaceutical composition
  • an aqueous pharmaceutical composition comprising the P2Y12 receptor antagonist 4-((R)-2- ⁇ [6-((S)-3-methoxy-pyrrolidin-1-yl)-2-phenyl- pyrimidine-4-carbonyl]-amino ⁇ -3-phosphono-propionyl)-piperazine-1-carboxylic acid butyl ester (hereinafter COMPOUND, also known as selatogrel) and a pharmaceutically acceptable buffer; and its use as a medicament by parenteral administration (especially subcutaneous (s.c.) or intradermal administration).
  • COMPOUND also known as selatogrel
  • COMPOUND The preparation and the medical use of COMPOUND is described in WO 2009/069100; WO 2018/055016; WO 2018/167139; Baldoni D et al., Clin Drug Investig (2014), 34(11), 807-818; Caroff E et al., J. Med. Chem. (2015), 58, 9133-9153; Storey R F et al., European Heart Journal, ehz807, doi:10.1093/eurheartj/ehz807; and Sinnaeve P R et al, J Am Coll Cardiol (2020), 75 (20), 2588-97 (doi.org/10.1016/j.jacc.2020.03.059).
  • the ADP receptors P2Yi and P2YI 2 play a critical role in platelet activation and aggregation (Andre P et al. (2003) J Clin Invest 112:398-406). Inhibition of the P2YI 2 receptor is a validated concept for prevention of major adverse cardiovascular events in patients with acute coronary syndromes (ACS) as demonstrated by the thienopyridines ticlopidine, clopidogrel and prasugrel (Franchi F et al. (2015) Nat Rev Cardiol 12:30-47). These drugs following metabolic activation irreversibly block the P2YI 2 receptor and platelet function (Antman EM et al. (2004) Circulation 110:588-636; Anderson JL et al.
  • ACS acute coronary syndromes
  • ticagrelor achieved a wider therapeutic window in rat and dog thrombosis models as compared with clopidogrel (van Giezen JJ et al. (2009) Thromb Res 124:565-571 ; Becker RC et al. (2010) Thromb Haemost 103:535-544). It was argued that the reversibility of the binding of ticagrelor to P2YI 2 might account for this difference. In patients, ticagrelor achieved a higher extent of inhibition of ADP-induced platelet aggregation than clopidogrel (Husted S et al.
  • ticagrelor showed superior efficacy and no significant difference in the risk of major bleeding events to clopidogrel.
  • a significant increase in fatal intracranial bleedings and in major or minor bleedings according to the study criteria was reported for ticagrelor (Wallentin L et al. (2009) N Engl J Med 361 :1045-1057).
  • COMPOUND was described to cause significantly less blood loss than ticagrelor at equivalent antithrombotic efficacy in rat (Rey M et al. (2017) Pharma Res Per, 5(5), e00338 (doi: 10.1002/prp2.338)).
  • the pharmaceutical composition comprising COMPOUND needs to be sufficiently stable for at least 12 months (especially at least 18 months and notably at least 24 months) if stored at about room temperature. Unfortunately, it was found that solutions of COMPOUND in water at lower pH values are not sufficiently stable.
  • COMPOUND 32 mg/ml_
  • Fig. 1 shows a Scanning Electron Microscopy (SEM) image of the inner part of the barrel of a glass syringe in two different magnifications.
  • the displayed area of the inner part of the barrel was in contact to an aqueous pharmaceutical composition comprising a boric acid buffer for 6 days at 70°C.
  • Fig. 2 shows a Scanning Electron Microscopy (SEM) image of the inner part of the barrel of a glass syringe in two different magnifications.
  • the displayed area of the inner part of the barrel was outside of the area that was in contact to an aqueous pharmaceutical composition comprising a boric acid buffer for 6 days at 70°C.
  • a first embodiment relates to an aqueous pharmaceutical composition
  • an aqueous pharmaceutical composition comprising: • 4-((R)-2- ⁇ [6-((S)-3-methoxy-pyrrolidin-1-yl)-2-phenyl-pyrimidine-4-carbonyl]- amino ⁇ -3-phosphono-propionyl)-piperazine-1 -carboxylic acid butyl ester
  • COMPOUND may be used in the preparation of the aqueous pharmaceutical composition in any form.
  • COMPOUND may be used in any amorphous or crystalline form, in anhydrous form or in form of a hydrate or a pharmaceutically acceptable solvate, in form of the free compound (i.e. non-salt form), in form of a pharmaceutically acceptable salt or in any mixture of such forms.
  • COMPOUND is used in the preparation of the aqueous pharmaceutical composition in form of the hydrochloride salt (especially in a polymorphic form disclosed in WO 2018/055016).
  • COMPOUND is used in the preparation of the aqueous pharmaceutical composition in form of the di-sodium salt.
  • COMPOUND is used in the preparation of the aqueous pharmaceutical composition in form of the free compound (non-salt form). It is preferred that COMPOUND, or a pharmaceutically acceptable salt thereof, is used in the preparation of the present pharmaceutical compositions in essentially pure form. The amount of COMPOUND may be adjusted taking into account the actual chemical purity, or the presence of a salt form and/or of a solvate or hydrate, of COMPOUND. It is further to be understood that the pH value of the aqueous pharmaceutical composition may be adjusted to the target value with a pharmaceutically acceptable base.
  • the pharmaceutically acceptable base used in the preparation of the aqueous pharmaceutical composition may be a pharmaceutically acceptable strong base, a pharmaceutically acceptable weak base different from the basic component of the pharmaceutically acceptable buffer, or the basic component of the pharmaceutically acceptable buffer that is used in the aqueous pharmaceutical composition.
  • the basic component of the pharmaceutically acceptable buffer may be in essentially pure form or in a mixture with the acidic component of the pharmaceutically acceptable buffer, wherein the ratio between the basic and the acidic component is sufficiently high to adjust the pH value of the aqueous pharmaceutical composition to the target value.
  • the pharmaceutically acceptable base is a pharmaceutically acceptable strong base, or the basic component of the pharmaceutically acceptable buffer that is used in the aqueous pharmaceutical composition.
  • the pharmaceutically acceptable base is a pharmaceutically acceptable strong base (especially sodium hydroxide).
  • the pharmaceutically acceptable base may be used in essentially pure form without a solvent, or as a solution in a pharmaceutically acceptable solvent or solvent mixture (especially a solution in water).
  • a solution of sodium hydroxide in water is used as the pharmaceutically acceptable base.
  • a pharmaceutically acceptable acid especially a pharmaceutically acceptable strong acid, such as aqueous hydrochloride acid, may be used for pH adjustment.
  • an aqueous pharmaceutical composition may change over time, especially if stored for one or several years.
  • an aqueous pharmaceutical composition is described and/or claimed to have a pH value between a specific lower limit and a specific upper limit, this means that the pH value of the aqueous pharmaceutical composition is in the given range defined by the specific lower limit and the specific upper limit at least immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • the pH value remains in the given range defined by the specific lower limit and the specific upper limit for at least 6 months (preferably at least 12 months and most preferably at least 18 months) after final preparation of the aqueous pharmaceutical composition.
  • pharmaceutically acceptable buffer refers to a mixture of a weak Bransted base (the basic component of the pharmaceutically acceptable buffer) and its conjugated acid (the acidic component of the pharmaceutically acceptable buffer), wherein both, the weak base and the conjugated acid exhibit minimal undesired toxicological effects.
  • the weak base may be an inorganic or organic base and may be in the form of a neutral molecule or in form of a salt comprising a protonatable anion.
  • boric acid is understood to be a weak Bransted acid and a boric acid buffer is thus comprised in the definition of a pharmaceutically acceptable buffer.
  • Examples of pharmaceutically acceptable buffer are ammonium buffer, boric acid buffer, carbonate buffer, phosphate buffer, arginine buffer, glycine buffer, histidine buffer, meglumine buffer (also known as N-methyl-D-glucamine buffer), 2-aminoethanol buffer (also known as monoethanolamine buffer), bis(2- hydroxyethyl)amine buffer (also known as diethanolamine buffer), and tris(hydroxymethyl)aminomethane buffer (also known as tromethamine or Tris buffer). It is preferred that the pharmaceutically acceptable buffer is different from phosphate buffer. Most preferred are boric acid buffer and arginine buffer.
  • the pharmaceutically acceptable buffer may be obtained from: ammonium chloride, ammonium bromide or ammonia (e.g. as solution in water) for ammonium buffer; boric acid or sodium tetraborate for boric acid buffer; sodium hydrogencarbonate or sodium carbonate for carbonate buffer; sodium dihydrogenphosphate or disodium hydrogenphosphate for phosphate buffer; arginine or arginine hydrochloride for arginine buffer; glycine or glycine hydrochloride for glycine buffer; histidine or histidine hydrochloride for histidine buffer; N-methyl-D-glucamine or N-methyl-D-glucamine hydrochloride for meglumine buffer; 2-aminoethanol or 2-aminoethanol hydrochloride for 2-aminoethanol buffer; bis(2-hydroxyethyl)amine or bis(2-hydroxyethyl)amine hydrochloride for bis(2-hydroxyethyl)amine buffer; tris(hydroxymethyl)aminomethane or tris(hydroxy
  • any reference to one of the amino acids arginine or histidine refers to the respective amino acid in its D-form, its L-form or any mixture (including a racemic mixture) thereof; and especially to the respective amino acid in its enantiomerically essentially pure L-form.
  • salts refers to salts that retain the desired biological activity of the subject compound and exhibit minimal undesired toxicological effects.
  • Such salts include inorganic or organic acid and/or base addition salts depending on the presence of basic and/or acidic groups in the subject compound.
  • “Handbook of Pharmaceutical Salts. Properties, Selection and Use.’ P. Heinrich Stahl, Camille G. Wermuth (Eds.), Wiley-VCH, 2008 and ‘Pharmaceutical Salts and Co-crystals’, Johan Wouters and Luc Quere (Eds.), RSC Publishing, 2012.
  • a preferred pharmaceutically acceptable salt of COMPOUND is the hydrochloride salt of COMPOUND (acid addition salt).
  • Other preferred pharmaceutically acceptable salts of COMPOUND are the alkali metal and alkaline earth metal salts of COMPOUND (base addition salts); especially the sodium and potassium salts and notably the sodium salts of COMPOUND. It is understood that in base addition salts one or both acidic protons of COMPOUND may be replaced by cations of the base.
  • the most preferred base addition salt of COMPOUND is the disodium salt of COMPOUND.
  • the term “pharmaceutically acceptable base” refers to a Bransted base whose reaction products from the adjustment of the pH value (e.g. a sodium salt and water if sodium hydroxide is used as base) exhibit minimal undesired toxicological effects.
  • pharmaceutically acceptable strong base refers to a “pharmaceutically acceptable base” that is completely or almost completely ionized or dissociated if dissolved in water. Examples of pharmaceutically acceptable strong bases are hydroxide salts of alkali or alkaline earth metals; especially sodium hydroxide or potassium hydroxide; and notably sodium hydroxide.
  • a solvate of COMPOUND is a “pharmaceutically acceptable solvate” if the solvent forming the solvate exhibits minimal undesired toxicological effects.
  • target value in relation to the pH value of the aqueous pharmaceutical composition (or alternatively “targeted pH value”) means the pH value to which the aqueous pharmaceutical composition should be adjusted.
  • target value in relation to the pH value of the aqueous pharmaceutical composition immediately after its final preparation is equal to the target value.
  • the term “essentially”, for example when used in a term such as "essentially pure” is understood in the context of the present invention to mean especially that the respective composition / component / compound / stereoisomer etc. consists in an amount of at least 90, especially of at least 95, and notably of at least 99 per cent by weight of the respective pure composition / component / compound / stereoisomer etc..
  • the term “consisting essentially of” is understood in the context of the present invention to mean that the respective composition consists in an amount of at least 90, especially of at least 95, notably of at least 99, and in particular in an amount of 100 per cent by weight (i.e. in the meaning of "consisting of) of the respective composition in the amounts as explicitly stated in the respective embodiment.
  • the total weight percent of the aqueous pharmaceutical composition as defined in embodiment 1), and embodiments 2) to 60) below is 100.
  • compositions as defined in embodiment 1) and 2) to 6) and 10) to 60) may additionally comprise further conventional ingredients and/ or additives, which may be used alone or in combination.
  • further conventional ingredients and/ or additives which may be used alone or in combination.
  • tonicity modifiers such as salts (e.g. NaCI, KCI, MgCI 2 , CaCI 2 ).
  • Further conventional ingredients or additives are for example antimicrobial preservatives such as used e.g. in bacteriostatic water for injection.
  • An example is benzyl alcohol.
  • the aqueous pharmaceutical composition may in addition to water further comprise up to 20 ww% (especially up to 10 ww% and notably up to 5 ww%) of a pharmaceutically acceptable solvent or a mixture of pharmaceutically acceptable solvents.
  • pharmaceutically acceptable solvent refers to a solvent that exhibits minimal undesired toxicological effects.
  • Representative examples of pharmaceutically acceptable solvents are polyethylene glycol, polysorbate, DMSO and NMP.
  • the aqueous pharmaceutical composition does not contain a pharmaceutically acceptable solvent in addition to water (other than trace amounts of residual solvent present in an ingredient used in the preparation of the aqueous pharmaceutical composition).
  • composition is interchangeable with the term “formulation”.
  • the term “about” placed before a numerical value “X” refers in the current application to an interval extending from X minus 10% of X to X plus 10% of X, and preferably to an interval extending from X minus 5% of X to X plus 5% of X (wherein it is well understood that the lower limit is equal or above 0% and the higher limit is equal or below 100%).
  • the term “about” placed before a temperature ⁇ ” refers in the current application to an interval extending from the temperature Y minus 10 °C to Y plus 10 °C; and preferably to an interval extending from Y minus 5 °C to Y plus 5 °C.
  • ww% refers to a percentage by weight compared to the total weight of the composition considered. If not explicitly stated otherwise, the considered total weight is the total weight of the aqueous pharmaceutical composition. In case a certain value is given as % value, in absence of further specification such value refers to ww%.
  • the expression (wt/wt) relating to a ratio refers to a ratio by weight of the respective components.
  • the invention relates to an aqueous pharmaceutical composition
  • an aqueous pharmaceutical composition comprising:
  • aqueous pharmaceutical composition has a pH value between 8.5 and 11.0.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) or 2), wherein the sum of the amount of COMPOUND, of the amount of the pharmaceutically acceptable buffer and of the amount of water is at least 80 ww% of the aqueous pharmaceutical composition.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) or 2), wherein the sum of the amount of COMPOUND, of the amount of the pharmaceutically acceptable buffer and of the amount of water is at least 90 ww% of the aqueous pharmaceutical composition.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) or 2), wherein the sum of the amount of COMPOUND, of the amount of the pharmaceutically acceptable buffer and of the amount of water is at least 95 ww% of the aqueous pharmaceutical composition.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) or 2), wherein the sum of the amount of COMPOUND, of the amount of the pharmaceutically acceptable buffer and of the amount of water is at least 98 ww% of the aqueous pharmaceutical composition.
  • the invention relates to an aqueous pharmaceutical composition according to embodiment 1), consisting essentially of:
  • COMPOUND in an amount between 0.1 to 20 ww%; a pharmaceutically acceptable buffer; • water; and
  • aqueous pharmaceutical composition has a pH value between 8.5 and
  • an aqueous pharmaceutical composition consisting essentially of:
  • a pharmaceutically acceptable buffer selected from a boric acid buffer or an arginine buffer; ⁇ water; and
  • aqueous pharmaceutical composition has a pH value between 8.7 and 9.5.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 6), wherein the pharmaceutical composition is obtainable by:
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) or 3) to 6), wherein the amount of COMPOUND is between 0.1 ww% and 20 ww%.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 9), wherein the amount of COMPOUND is between 0.7 ww% and 7.0 ww%.
  • Lower limits of the amount of COMPOUND are 0.7 ww%, 1.0 ww% and 2.0 ww%, upper limits are 7.0 ww%, 5.0 ww% and 4.0 ww%. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • a preferred amount of COMPOUND is from 1.0 ww% to 5.0 ww%.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 9), wherein the amount of COMPOUND is between 2.4 ww% and 3.8 ww%.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) or 3) to 6), wherein the mass concentration of COMPOUND in the aqueous pharmaceutical composition is between 6 mg/mL and 60 mg/mL, preferably between 15 mg/mL and 45 mg/mL and most preferably between 25 mg/mL and 40 mg/mL.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 9), wherein the mass concentration of COMPOUND in the aqueous pharmaceutical composition is about 32 mg/mL.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 7) or 10) to 14), wherein the pharmaceutically acceptable buffer has a pKa value in water at 25°C between 8.0 and 11.0.
  • Lower limits of the pKa value are 8.0, 8.5 and 8.8, upper limits are 11.0, 10.0 and 9.5. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed. It is further to be understood that in the case of a di- or triprotic acid only one of the two or three pKa values has to be in the above-mentioned range.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 7) or 10) to 14), wherein the pharmaceutically acceptable buffer has a pKa value in water at 25°C between 8.8 and 9.5.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 7) or 10) to 14), wherein the pharmaceutically acceptable buffer is selected from ammonium buffer, boric acid buffer, carbonate buffer, phosphate buffer, arginine buffer, glycine buffer, histidine buffer, meglumine buffer, or tris(hydroxymethyl)aminomethane buffer (preferably from ammonium buffer, boric acid buffer, carbonate buffer, arginine buffer, glycine buffer, histidine buffer, meglumine buffer, or tris(hydroxymethyl)aminomethane buffer).
  • the pharmaceutically acceptable buffer is selected from ammonium buffer, boric acid buffer, carbonate buffer, phosphate buffer, arginine buffer, glycine buffer, histidine buffer, meglumine buffer, or tris(hydroxymethyl)aminomethane buffer.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 14), wherein the pharmaceutically acceptable buffer is selected from boric acid buffer, or arginine buffer (preferably boric acid buffer, or L-arginine buffer).
  • the pharmaceutically acceptable buffer is selected from boric acid buffer, or arginine buffer (preferably boric acid buffer, or L-arginine buffer).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 14), wherein the pharmaceutically acceptable buffer is boric acid buffer.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 14), wherein the pharmaceutically acceptable buffer is arginine buffer.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 14), wherein the pharmaceutically acceptable buffer is L-arginine buffer.
  • L-arginine buffer refers to an arginine buffer wherein the enantiomeric ratio between the L-form and the D-form is at least 90/10, preferably at least 95/5, more preferably at least 99/1 , and most preferably at least 99.9/0.1.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 21), wherein the molar concentration of the pharmaceutically acceptable buffer in the aqueous pharmaceutical composition is between 10 mmol/L and 300 mmol/L.
  • Lower limits of the molar concentration of the pharmaceutically acceptable buffer in the aqueous pharmaceutical composition are 10 mmol/L, 25 mmol/L, and 40 mmol/L
  • upper limits are 300 mmol/L, 200 mmol/L, and 100 mmol/L. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • molar concentration of the pharmaceutically acceptable buffer refers to the sum of the molar concentrations of all different protonated and deprotonated components of the buffer system in the respective aqueous pharmaceutical composition.
  • the “molar concentration of the pharmaceutically acceptable buffer” may be calculated by dividing the amount of the added buffer component during preparation of the aqueous pharmaceutical composition (in moles) by the total volume of the finally prepared aqueous pharmaceutical composition (in litres).
  • the term “molar concentration of the pharmaceutically acceptable buffer” refers to the sum of the molar concentration of the basic component of the pharmaceutically acceptable buffer and of the molar concentration of the acidic component of the pharmaceutically acceptable buffer in the respective aqueous pharmaceutical composition.
  • the buffer capacity of the pharmaceutically acceptable buffer in the aqueous pharmaceutical composition is between 10 mM/pH and 170 mM/pH.
  • Lower limits of the buffer capacity are 10 mM/pH, 15 mM/pH, and 20 mM/pH; upper limits are 170 mM/pH, 100 mM/pH, and 60 mM/pH. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the buffer capacity is between 20 mM/pH and 60 mM/pH.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 21), wherein the molar concentration of the pharmaceutically acceptable buffer in the aqueous pharmaceutical composition is between 25 mmol/L and 200 mmol/L.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 21), wherein the molar concentration of the pharmaceutically acceptable buffer in the aqueous pharmaceutical composition is between 40 mmol/L and 100 mmol/L.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 21), wherein the molar concentration of the pharmaceutically acceptable buffer in the aqueous pharmaceutical composition is about 60 mmol/L.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 25), wherein the water is water for injection (WFI), sterile water for injection (SWFI), or bacteriostatic water for injection (BWFI).
  • WFI water for injection
  • SWFI sterile water for injection
  • BWFI bacteriostatic water for injection
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 25), wherein the water is water for injection (WFI).
  • WFI water for injection
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 7) or 9) to 27), wherein the aqueous pharmaceutical composition has a pH value between 8.5 and 11.0.
  • Lower limits of the pH value of the aqueous pharmaceutical composition are 8.5, 8.7, and 8.9; upper limits are 11.0, 9.5, and 9.3. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 27), wherein the aqueous pharmaceutical composition has a pH value between 8.7 and 9.5.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 27), wherein the aqueous pharmaceutical composition has a pH value between 8.9 and 9.3.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 30), wherein the inorganic salt, if present, is selected from salts of formula MX, wherein M represents lithium, sodium, or potassium and X represents chlorine or bromine; or MX 2 , wherein M represents magnesium or calcium and X represents chlorine or bromine.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 30), wherein the inorganic salt, if present, is selected from sodium chloride, potassium chloride, magnesium chloride, calcium chloride, or any mixture thereof (and preferably sodium chloride, potassium chloride, or any mixture thereof; and most preferably sodium chloride).
  • the inorganic salt if present, is selected from sodium chloride, potassium chloride, magnesium chloride, calcium chloride, or any mixture thereof (and preferably sodium chloride, potassium chloride, or any mixture thereof; and most preferably sodium chloride).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 30), wherein the aqueous pharmaceutical composition comprises sodium chloride.
  • the inorganic salt (especially sodium chloride) is used to adjust the osmolality of the aqueous pharmaceutical composition to a value > 230 mOsm/kg and notably to a value between 230 mOsm/kg and 1000 mOsm/kg.
  • Lower limits of the osmolality of the aqueous pharmaceutical composition are 230 mOsm/kg, 250 mOsm/kg, and 270 mOsm/kg; upper limits are 1000 mOsm/kg, 500 mOsm/kg, and 330 mOsm/kg. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the osmolality is between 270 mOsm/kg and 330 mOsm/kg.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 33), wherein the aqueous pharmaceutical composition is in a container.
  • the invention relates to an aqueous pharmaceutical composition according to embodiment 34), wherein the container is selected from a vial, an ampoule, a cartridge, or a syringe.
  • the invention relates to an aqueous pharmaceutical composition according to embodiment 34), wherein the container is a syringe.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 34) to 36), wherein the container is a glass container (preferably a glass syringe).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 34) to 37), wherein the container has an inner volume between 0.1 mL and 5.0 mL.
  • Lower limits of the inner volume are 0.1 mL, 0.3 mL, 0.5 mL and 0.8 mL, upper limits are 5.0 mL, 3.0 mL, 2.3 mL and 1.0 mL. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 34) to 37), wherein the container has an inner volume between 0.5 mL and 2.3 mL.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 34) to 39), wherein the amount of the aqueous pharmaceutical composition in the container corresponds to the amount of the unit dose.
  • unit dose means the amount of the aqueous pharmaceutical composition that is administered and/or is to be administered to a patient in a single dose.
  • aqueous pharmaceutical composition when used in the context of “corresponds to the amount of the unit dose”, means that the amount of the aqueous pharmaceutical composition equals the amount of the unit dose plus a small addition to compensate for a loss of aqueous pharmaceutical composition during administration, such loss being caused for instance by the dead volume of the respective injection device (e.g. a syringe, an autoinjector, a medical pump or the like).
  • the respective injection device e.g. a syringe, an autoinjector, a medical pump or the like.
  • the invention relates to an aqueous pharmaceutical composition according to embodiment 40), wherein the volume of the aqueous pharmaceutical composition in the container is between 0.1 mL and 3.0 mL.
  • Lower limits of the volume of the aqueous pharmaceutical composition in the container are 0.1 mL, 0.2 mL, 0.3 mL and 0.4 mL, upper limits are 3.0 mL, 1.5 mL, 1.0 mL and 0.7 mL. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the invention relates to an aqueous pharmaceutical composition according to embodiment 40), wherein the volume of the aqueous pharmaceutical composition in the container is between 0.3 mL and 1 .0 mL (preferably between 0.4 mL and 0.7 mL).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 40) to 42), wherein the amount of COMPOUND in the aqueous pharmaceutical composition in the container is between 4.0 mg and 32.0 mg.
  • Lower limits of the amount of COMPOUND in the aqueous pharmaceutical composition in the container are 4.0 mg, 7.0 mg, 10.0 mg and 14.0 mg, upper limits are 32.0 mg, 28.0 mg, 24.0 mg and 20.0 mg. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 40) to 42), wherein the amount of COMPOUND in the aqueous pharmaceutical composition in the container is between 14.0 mg and 20.0 mg.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiment 34) to 39), wherein the amount of the aqueous pharmaceutical composition in the container corresponds to two or more (up to a maximum of ten) unit doses (preferably two to five and most preferably two or three unit doses).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 34) to 45), wherein the container is inserted in an autoinjector device (preferably a pen-injector device).
  • an autoinjector device preferably a pen-injector device
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 46), wherein the aqueous pharmaceutical composition is an emulsion, a suspension or a solution.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 46), wherein the aqueous pharmaceutical composition is a solution.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 48), wherein the aqueous pharmaceutical composition is sterile.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 49), wherein the aqueous pharmaceutical composition is pyrogen-free.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 50), wherein the amount of COMPOUND in the aqueous pharmaceutical composition after storage for 1 year at 25°C is at least 80% (preferably at least 85% and most preferably at least 90%) of the amount of COMPOUND in the aqueous pharmaceutical composition immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 50), wherein the amount of COMPOUND in the aqueous pharmaceutical composition after storage for 2 years at 25°C is at least 80% (preferably at least 85% and most preferably at least 90%) of the amount of COMPOUND in the aqueous pharmaceutical composition immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 50), wherein the amount of COMPOUND in the aqueous pharmaceutical composition after storage for 3 years at 25°C is at least 80% (preferably at least 85% and most preferably at least 90%) of the amount of COMPOUND in the aqueous pharmaceutical composition immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 50), wherein the amount of COMPOUND in the aqueous pharmaceutical composition after storage for 18 months at 30°C is at least 80% (preferably at least 85% and most preferably at least 90%) of the amount of COMPOUND in the aqueous pharmaceutical composition immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 50), wherein the amount of COMPOUND in the aqueous pharmaceutical composition after storage for 2 years at 30°C is at least 80% (preferably at least 85% and most preferably at least 90%) of the amount of COMPOUND in the aqueous pharmaceutical composition immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 50), wherein the amount of COMPOUND in the aqueous pharmaceutical composition after storage for 6 month at 2°C to 8°C (especially 5°C) and subsequently for 2 years at 25°C is at least 80% (preferably at least 85% and most preferably at least 90%) of the amount of COMPOUND in the aqueous pharmaceutical composition immediately (for instance 1 hour or 2 hours) after final preparation of the aqueous pharmaceutical composition (i.e. after all ingredients are added to the aqueous pharmaceutical composition).
  • COMPOUND and/or the main degradation product of COMPOUND ((R)-2-(6-((S)-3-methoxypyrrolidin-1-yl)-2-phenylpyrimidine-4-carboxamido)-3- phosphonopropanoic acid) can form covalently bound adducts to the pharmaceutically acceptable buffer used in the aqueous pharmaceutical composition.
  • Such adducts are disadvantageous, inter alia as their toxicological profiles are unknown. Thus, additional toxicological studies may be required, or toxicologically relevant findings may be identified. For instance, the U.S.
  • FDA Food and Drug Administration
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 56), wherein the ratio between (i) the amount of a covalently bound adduct of the pharmaceutically acceptable buffer to COMPOUND and (ii) the amount of COMPOUND in the aqueous pharmaceutical composition is less than 1 to 250 (preferably less than 1 to 500) after storage of the aqueous pharmaceutical composition for 2 years at 25°C.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 56), wherein the ratio between (i) the amount of a covalently bound adduct of the pharmaceutically acceptable buffer to ((R)-2- ⁇ [6-((S)-3-methoxy-pyrrolidin-1-yl)-2-phenyl-pyrimidine-4-carbonyl]-amino ⁇ -3- phosphono-propanoic acid and (ii) the amount of COMPOUND in the aqueous pharmaceutical composition is less than 1 to 250 (preferably less than 1 to 500) after storage of the aqueous pharmaceutical composition for 2 years at 25°C.
  • the term “amount” refers to the number of molecules, wherein the number of molecules may be given in any unit directly related to the number of molecules (such as “mol”).
  • the aqueous pharmaceutical composition may modify the surface of glass. This modification may be caused by glass corrosion and/or delamination and results in an increased roughness of the glass surface and/or the occurrence of particles in the aqueous pharmaceutical composition, especially after storage of the aqueous pharmaceutical composition in a glass container such as a glass syringe.
  • the increased roughness and/or the occurrence of particles has several disadvantages. For instance, in case a glass syringe is used for storage of the aqueous pharmaceutical composition and (after storage) for administration to a patient, the occurrence of particles may result in a risk for the patient if such glass particles are injected to the patient (especially if accidentally injected into a vein).
  • An increased roughness of the glass surface of the inner wall of the syringe may result in an increase of the force that is needed to move the plunger/piston within the barrel of the syringe.
  • an autoinjector device such as a pen-injector device
  • this may result in a malfunction of the autoinjector device.
  • boric acid buffer resulted in a significant higher glass corrosion and/or delamination.
  • the invention relates to an aqueous pharmaceutical composition according to any one of embodiments 1) to 58), wherein the aqueous pharmaceutical composition comprises less than 10000 (preferably less than 3000, more preferably less than 500) particles with a particle size > 10pm per millilitre of the aqueous pharmaceutical composition after storage of the aqueous pharmaceutical composition at 70°C for 6 days in a glass syringe.
  • the amount and size of the particles may be measured for instance using micro flow imaging (MFI) (alternatively called flow imaging microscopy (FIM) or dynamic imaging analysis (DIA)) as described in the experimental part.
  • MFI micro flow imaging
  • FIM flow imaging microscopy
  • DIA dynamic imaging analysis
  • the invention relates to an aqueous pharmaceutical composition according to embodiment 59), wherein the glass syringe is a siliconized glass syringe.
  • the silicone is covalently bound to the inner surface of the glass barrel, for instance by plasma treatment.
  • the invention in another embodiment, relates to a container comprising (especially containing) an aqueous pharmaceutical composition according to any one of embodiments 1) to 60).
  • the invention in another embodiment, relates to a container according to embodiment 61), wherein the container is selected from a vial, an ampoule, a cartridge, or a syringe.
  • the invention in another embodiment, relates to a container according to embodiment 61), wherein the container is a syringe.
  • the invention relates to a container according to any one of embodiment 61) to 63), wherein the container is a glass container (preferably a glass syringe).
  • the invention relates to a container according to any one of embodiment 61) to 64), wherein the container has an inner volume between 0.1 mL and 5.0 mL.
  • Lower limits of the inner volume are 0.1 mL, 0.3 mL, 0.5 mL and 0.8 mL, upper limits are 5.0 mL, 3.0 mL, 2.3 mL and 1.0 mL. It is to be understood that each lower limit can be combined with each upper limit. Hence all combinations shall herewith be disclosed.
  • the invention relates to a container according to any one of embodiment 61) to 64), wherein the container has an inner volume between 0.5 mL and 2.3 mL.
  • the invention relates to a container according to any one of embodiment 61) to 66) for use in an autoinjector device (especially a pen-injector device).
  • a further embodiment relates to a method for the preparation of an aqueous pharmaceutical composition; said method comprising the steps of: a) dissolving the pharmaceutically acceptable buffer (i.e. the basic component of the pharmaceutically acceptable buffer, the acidic component of the pharmaceutically acceptable buffer or any mixture thereof) and COMPOUND, or a pharmaceutically acceptable salt thereof, in water; and b) adjusting the pH value of the aqueous pharmaceutical composition to a pH value between 8.2 and 12.0.
  • the pharmaceutically acceptable buffer i.e. the basic component of the pharmaceutically acceptable buffer, the acidic component of the pharmaceutically acceptable buffer or any mixture thereof
  • COMPOUND or a pharmaceutically acceptable salt thereof
  • a further embodiment relates to a method according to embodiment 68), wherein the pharmaceutically acceptable buffer is selected from arginine buffer or boric acid buffer.
  • a further embodiment relates to a method according to any one of embodiments 68) or 69), wherein water is water for injection.
  • a further embodiment relates to a method according to any one of embodiments 68) to 70), wherein the pH value is between 8.5 and 11.0.
  • a further embodiment relates to a method according to any one of embodiments 68) to 70), wherein the pH value is between 8.7 and 9.5.
  • a further embodiment relates to a method according to any one of embodiments 68) to 70), wherein the pH value is between 8.9 and 9.3.
  • a further embodiment relates to a method according to any one of embodiments 68) to 73), wherein the pH value is adjusted with a pharmaceutically acceptable strong base (and especially with an aqueous solution of sodium hydroxide).
  • a further embodiment relates to a method for the preparation of container according to any of embodiments 61) to 67), wherein the aqueous pharmaceutical composition is filled in the container using aseptic processing.
  • aseptic processing refers to the sterile filtration and subsequent filling of a pharmaceutical composition (especially an aqueous pharmaceutical composition) into a sterilized container under sterile conditions.
  • a pharmaceutical composition especially an aqueous pharmaceutical composition
  • the advantage of an aseptic processing is the avoidance of thermal stress of the aqueous pharmaceutical composition and its ingredients due to a thermal sterilization (such as for instance a steam sterilization) of the finally filled container.
  • the aqueous pharmaceutical composition according to the invention may be used as a medicament.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of a disease or disorder, wherein the disease or disorder is selected from acute arterial thromboses or acute venous thromboses.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of a disease or disorder, wherein the disease or disorder is selected from acute arterial thromboses.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of a disease or disorder, wherein the disease or disorder is selected from acute coronary syndromes, percutaneous intervention (PCI) complications, stent thrombosis, periprocedural thrombotic events, myocardial infarction (especially acute myocardial infarction), peripheral ischaemia, amaurosis, sudden cardiac death, ischaemic stroke or transient ischaemic attack.
  • PCI percutaneous intervention
  • stent thrombosis periprocedural thrombotic events
  • myocardial infarction especially acute myocardial infarction
  • peripheral ischaemia amaurosis
  • sudden cardiac death ischaemic stroke or transient ischaemic attack.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of a disease or disorder, wherein the disease or disorder is selected from acute coronary syndromes, myocardial infarction (especially acute myocardial infarction), peripheral ischaemia, amaurosis, ischaemic stroke or transient ischaemic attack.
  • a disease or disorder is selected from acute coronary syndromes, myocardial infarction (especially acute myocardial infarction), peripheral ischaemia, amaurosis, ischaemic stroke or transient ischaemic attack.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of acute coronary syndromes.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of myocardial infarction.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of acute myocardial infarction.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of peripheral ischaemia.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of amaurosis.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis of sudden cardiac death.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment (especially treatment) of a disease, wherein the disease is selected from ischaemic stroke or transient ischaemic attack.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the emergency treatment of suspected acute coronary syndromes (ACS) or suspected acute myocardial infarction (AMI) by patient self-administration prior to hospitalization.
  • ACS suspected acute coronary syndromes
  • AMD suspected acute myocardial infarction
  • Acute coronary syndromes refers to syndromes due to sudden decrease or interruption of blood flow in some coronary arteries.
  • Acute coronary syndromes encompass ST elevation myocardial infarction (STEMI), non ST elevation myocardial infarction (NSTEMI) and unstable angina. It is understood that an aqueous pharmaceutical composition that is disclosed to be useful in the prevention or treatment of ACS is likewise useful in the prevention or treatment of STEMI, NSTEMI and/or unstable angina.
  • the term “emergency treatment of suspected acute coronary syndromes” or “emergency treatment of suspected acute myocardial infarction” refers to a treatment of a patient wherein the patient shows symptoms of ACS or AMI, respectively, such as suddenly occurring chest pain, chest discomfort (intermittent or not), persistent retrosternal pressure or heaviness radiating to the left arm, neck, back or jaw lasting for at least 10 min, nausea/vomiting, shortness of breath, fatigue, palpitations, lightheadedness or syncope (and especially clear symptoms of ACS/AMI such as suddenly occurring chest pain, chest discomfort (intermittent or not), or persistent retrosternal pressure or heaviness radiating to the left arm, neck, back or jaw lasting for at least 10 min); and wherein the patient is to be treated and/or requires treatment before an electrocardiogram, a chest X-ray and/or blood tests could be performed.
  • symptoms of ACS or AMI respectively, such as suddenly occurring chest pain, chest discomfort (intermittent or not), persistent retrosternal pressure or
  • the patient is a patient who was already known to have a high risk to suffer from ACS/AMI before the symptoms (especially clear symptoms) of ACS/AMI occurred, such as for instance a patient with a known coronary artery disease who had a prior symptomatic episode of acute coronary syndromes/acute myocardial infarction.
  • the treatment is effected by patient self-administration before hospitalization; it is preferred that the patient has received a training by a health care professional to better assess the symptoms of ACS/AMI before any such self-administration.
  • CAD coronary artery disease
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the emergency treatment of suspected acute myocardial infarction (AMI) by patient selfadministration prior to hospitalization.
  • AMD suspected acute myocardial infarction
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment of a disease, wherein the prevention and/or prophylaxis of the formation of platelet thrombi is indicated and/or required.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the prevention/prophylaxis or treatment of a disease, wherein the accelerated dissolution of newly formed platelet thrombi is indicated and/or required.
  • the aqueous pharmaceutical composition according to the invention may be used for the preparation of a medicament, and / or is suitable, for use in the treatment of a disease, wherein the reduction of the size of existent platelet thrombi is indicated and/or required.
  • an aqueous pharmaceutical composition is described as useful for the preparation of a medicament for the prevention/prophylaxis or treatment of certain diseases or disorders, or as suitable for use in the prevention or treatment of certain diseases or disorders, such pharmaceutical composition is likewise suitable for use in a method of preventing or treating said diseases or disorders comprising administering to a subject (notably a mammal, especially a human) in need thereof a pharmaceutically active amount of said aqueous pharmaceutical composition.
  • the aqueous pharmaceutical composition according to the invention is suitable for parenteral administration (such as subcutaneous or intradermal administration); especially for subcutaneous administration; and notably for subcutaneous administration by patient self-administration using an autoinjector device (especially a pen-injector device). 92)
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered by parenteral administration.
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered by subcutaneous or intradermal administration.
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered by subcutaneous administration.
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered by a health care provider.
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered by patient self-administration.
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered in a prehospital treatment.
  • the aqueous pharmaceutical composition according to the invention is administered and/or is to be administered by patient self-administration using an autoinjector device (especially a pen-injector device) in a prehospital treatment.
  • an autoinjector device especially a pen-injector device
  • the chemical stability of the aqueous pharmaceutical composition may be tested in conventional manner, e.g. by measurement of COMPOUND and its degradation products (such as especially (R)-2-(6-((S)-3-methoxypyrrolidin-1-yl)-2-phenylpyrimidine-4- carboxamido)-3-phosphonopropanoic acid).
  • the amounts of COMPOUND and its degradation products in a sample may be for instance evaluated via reversed-phase HPLC.
  • the aqueous pharmaceutical composition according to any one of embodiments 1) to 60) may be formulated in containers.
  • a batch size corresponding to 320g of COMPOUND corresponding to 339g COMPOUND hydrochloride; batch size of 10.15 kg of aqueous pharmaceutical composition
  • COMPOUND hydrochloride the following amounts of ingredients may be used:
  • the amounts may be adjusted for the purity of the respective ingredient / material.
  • the amount of COMPOUND hydrochloride (16.9mg) corresponds to 16.0mg COMPOUND.
  • the amount of COMPOUND in the respective form may be adjusted such that the amount of COMPOUND in the unit dose is 16.0mg; in addition, in case another form of COMPOUND than COMPOUND hydrochloride is used, the above amounts of NaOH, NaCI and water for injection may be adjusted to compensate for a different amount of NaOH needed for pH adjustment.
  • the amount of COMPOUND in the unit dose may vary within the limits accepted by the health authorities and especially within a range of 16.0mg ⁇ 1.6mg; this variation may come along with a respective change in the ratio between the ingredients or with a respective change in the total amount of the unit dose or with a combination of both.
  • the aqueous pharmaceutical composition may contain the following concentrations of ingredients:
  • the process for the preparation of an aqueous pharmaceutical composition as described before and filled in a container according to the present invention may comprise the following steps: ⁇ add specific amount (e.g. 7kg) of WFI into the reactor;
  • Raw materials may be purchased from commercial suppliers, or may be prepared by methods known in the art. Sample preparation for High Performance Liquid Chromatography and method used
  • HPLC-UV method used HPLC system: High pressure mixing Shimadzu Nexera X2
  • Solvent A tris(hydroxymethyl)aminomethane buffer 50mM pH 9 (adjusted with HCI) + 5% (v/v) acetonitrile
  • Solvent B Acetonitrile + 5% (v/v) tris(hydroxymethyl)aminomethane buffer 50mM pH 9 (adjusted with HCI)
  • COMPOUND and its HCI salt may be prepared according to the procedures as disclosed in WO 2009/069100 (example 2), WO 2018/055016 or Caroff E et al., J. Med. Chem. (2015), 58, 9133-9153.
  • Example 1
  • COMPOUND and L-Arginine were weight into a 20ml_ volumetric flask. 10ml_ water for injection was added and the suspension stirred. This suspension was then titrated to pH 9.25 resulting in full dissolution. Prior to filling, the formulation was filtered through 0.22pm PVDF filter. Preparation of the primary packaging
  • 0.5 mL of the sterile filtered formulation was filled into a sterile ready-to-use pre-fillable glass-syringe. The filling takes place under sterile and particle free conditions.
  • Example 2 Preparation and composition of an aqueous pharmaceutical composition comprising a boric acid buffer:
  • Additional buffers are prepared at a fixed buffer capacity of 34 mM/pH at pH 9.25.
  • COMPOUND and the respective buffer were weight into 20ml_ volumetric flasks. 10 mL water for injection was added to each flask and the suspensions were stirred. These suspensions were then titrated to pH 9.25 which led to full dissolution. Prior to filling the formulations into 1ml_ sterile, pre-fillable glass-syringes they were filtered through 0.22pm PVDF filter.
  • Aqueous formulations containing 32 mg/ml_ (52mM) COMPOUND were titrated to pH 7.0, 8.0, and 9.0 using aqueous 1N NaOH solution.
  • Example 5 Stability of COMPOUND in aqueous solution at different pH values in the presence or absence of a buffer
  • Aqueous formulations containing 32 mg/ml_ (52mM) COMPOUND and 20mM KH2P04 were titrated to pH 7.0, 7.5, 8.0 using aqueous 1N NaOH solution.
  • Aqueous formulations containing 52mM COMPOUND and 20mM boric acid were titrated to pH 8.0, 8.5, 9.0, 9.5, 10.0 using aqueous 1N NaOH solution.
  • a formulation containing 52mM COMPOUND without added buffer was titrated to pH 9.0 using aqueous 1N NaOH solution.
  • Table 11 Content of COMPOUND after storage of aqueous formulations containing different boric acid buffer concentrations at 75°C in dependence of the storage time
  • the syringes filled with aqueous pharmaceutical compositions as described under example 1 (L-arginine buffer), example 2 (boric acid buffer) and example 3 were stored at 55.3°C for 19 days.
  • the content of COMPOUND and of the main degradation product (R)- 2-(6-((S)-3-methoxypyrrolidin-1-yl)-2-phenylpyrimidine-4-carboxamido)-3- phosphonopropanoic acid was determined by HPLC analysis using conditions as described above.
  • Example 2 32mg/ml_ COMPOUND in 60mM boric acid buffer, pH 9.25, filled in 1ml_ pre-fillable glass-syringes
  • Example 1 32mg/ml_ COMPOUND in 60mM Arginine buffer, pH 9.25, filled in 1ml_ pre-fillable glass-syringes
  • Micro-Flow Imaging uses a CCD camera to capture bright field images in a flow cell. It has an optical magnification and detects particles in a size range from 2.0 pm to approximately 70 pm. The particles are counted and imaged.
  • Table 13 Particle count and particle size distribution in aqueous formulations stored in glass-syringes for 6 days at 70°C cumulative particle counts
  • Fig. 1 shows the SEM images of an area of the inner part of the glass barrel that was in contact with the boric acid formulation.
  • Fig. 2 shows the SEM images of an area of the inner part of the glass barrel that was not in contact with the boric acid formulation.
  • test samples were prepared by filling 0.54 mL of the above solution into prefillable 1 mL glass syringes with stacked in needle and stored under the conditions given in table 14 and 15. The samples were analysed after the given timepoints by ultraperformance liquid chromatography (UPLC) using the following conditions:
  • Reference standard solution 0.32 mg/mL of COMPOUND reference standard in diluent.
  • Sample solution The samples were diluted to 0.32 mg/mL of COMPOUND (based on amount of COMPOUND in the initial solution) with diluent Column: 100mm x 3.0 mm, 3 pm C18 column (e.g. YMC Triart C18 ExRS) Detection wavelength: 246 nm Flow rate: 0.5 mL/min Gradient: Table 14: Amount of COMOUND in the test samples after storage for up to 12 months under the given conditions
  • Table 15 Amount of DEGRADATION COMPOUND ((R)-2-(6-((S)-3-methoxypyrrolidin-1- yl)-2-phenylpyrimidine-4-carboxamido)-3-phosphonopropanoic acid) in the test samples after storage for up to 12 months under the given conditions

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dermatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medical Preparation Storing Or Oral Administration Devices (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)

Abstract

La présente invention concerne une composition pharmaceutique aqueuse comprenant l'antagoniste du récepteur P2Y12 : ester butylique d'acide 4-((R)-2-{[6-((S)-3-méthoxy-pyrrolidin-1-yl)-2-phényl-pyrimidine-4-carbonyl]-amino}-3-phosphono-propionyl)-pipérazine-1-carboxylique et un tampon pharmaceutiquement acceptable et son utilisation en tant que médicament par administration parentérale.
EP21739730.6A 2020-07-15 2021-07-14 Composition pharmaceutique aqueuse comprenant un antagoniste du récepteur p2y12 Pending EP4181872A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP2020069976 2020-07-15
PCT/EP2021/069579 WO2022013276A1 (fr) 2020-07-15 2021-07-14 Composition pharmaceutique aqueuse comprenant un antagoniste du récepteur p2y12

Publications (1)

Publication Number Publication Date
EP4181872A1 true EP4181872A1 (fr) 2023-05-24

Family

ID=76845245

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21739730.6A Pending EP4181872A1 (fr) 2020-07-15 2021-07-14 Composition pharmaceutique aqueuse comprenant un antagoniste du récepteur p2y12

Country Status (10)

Country Link
US (1) US20230255987A1 (fr)
EP (1) EP4181872A1 (fr)
KR (1) KR20230041732A (fr)
CN (1) CN115803012A (fr)
AU (1) AU2021309018A1 (fr)
CA (1) CA3185564A1 (fr)
IL (1) IL299828A (fr)
MX (1) MX2023000601A (fr)
TW (1) TW202216150A (fr)
WO (1) WO2022013276A1 (fr)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2225253E (pt) 2007-11-29 2012-09-19 Actelion Pharmaceuticals Ltd Derivados de ácido fosfónico e respetiva utilização como antagonistas do recetor p2y12
MA46266B1 (fr) 2016-09-22 2022-02-28 Idorsia Pharmaceuticals Ltd Formes cristallines
CA3050831A1 (fr) 2017-03-15 2018-09-20 Idorsia Pharmaceuticals Ltd Administration sous-cutanee d'un antagoniste du recepteur p2y12

Also Published As

Publication number Publication date
AU2021309018A1 (en) 2023-03-16
CN115803012A (zh) 2023-03-14
KR20230041732A (ko) 2023-03-24
IL299828A (en) 2023-03-01
US20230255987A1 (en) 2023-08-17
WO2022013276A1 (fr) 2022-01-20
MX2023000601A (es) 2023-02-13
CA3185564A1 (fr) 2022-01-20
TW202216150A (zh) 2022-05-01
JP2023532816A (ja) 2023-07-31

Similar Documents

Publication Publication Date Title
AU2018229750B2 (en) Pharmaceutical composition comprising selexipag
JP6835831B2 (ja) 非プロトン性極性溶媒中の安定な治療用グルカゴン製剤を製造するための方法
WO2016059588A1 (fr) Composition injectable stable de bivalirudine et son procede de preparation
JP6516831B2 (ja) シクロホスファミド液状濃縮物の製剤
JP6736765B2 (ja) ベンダムスチン溶液製剤
AU2018234056B2 (en) Subcutaneous administration of a P2Y12 receptor antagonist
KR102375798B1 (ko) 세바코일 디날부핀 에스테르의 서방출을 위한 약제학적 제형
EP1205181B9 (fr) Composition et procédé pour l'administration parentérale de D,L- ou L-lysinate d'ibuprofène
EP2922530B1 (fr) Compositions d'acétate de caspofungine
US20230255987A1 (en) Aqueous pharmaceutical composition comprising a p2y12 receptor antagonist
JP7490881B2 (ja) P2y12受容体アンタゴニストを有する水性医薬組成物
WO2015174948A1 (fr) Formulations comprenant des composés de lipoyle
JP2014516946A (ja) エザチオスタットとの賦形剤適合性

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230215

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
RAV Requested validation state of the european patent: fee paid

Extension state: TN

Effective date: 20230215

Extension state: MA

Effective date: 20230215

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40094173

Country of ref document: HK